Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 157
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Physiol Rev ; 100(4): 1839-1850, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32721181

RESUMO

Cancer patients appear to be more likely to be diagnosed with coronavirus disease 2019 (COVID-19). This is supported by the understanding of immunometabolic pathways that intersect patients with infection and cancer. However, data derived by case series and retrospective studies do not offer a coherent interpretation, since data from China suggest an increased risk of COVID-19, while data from the United States and Italy show a prevalence of COVID-19 in cancer patients comparable with the general population. Noteworthy, cancer and COVID-19 exploit distinct patterns of macrophage activation that promote disease progression in the most severe forms. In particular, the alternative activation of M2-polarized macrophages plays a crucial role in cancer progression. In contrast, the macrophage-activation syndrome appears as the source of M1-related cytokine storm in severe COVID-19 disease, thus indicating macrophages as a source of distinct inflammatory states in the two diseases, nonetheless as a common therapeutic target. New evidence indicates that NAMPT/NAD metabolism can direct both innate immune cell effector functions and the homeostatic robustness, in both cancer and infection. Moreover, a bidirectional relationship exists between the metabolism of NAD and the protective role that angiotensin converting enzyme 2, the COVID-19 receptor, can play against hyperinflammation. Within this immunometabolic framework, the review considers possible interference mechanisms that viral infections and tumors elicit on therapies and provides an overview for the management of patients with cancer affected by COVID-19, particularly for the balance of risk and benefit when planning normally routine cancer treatments and follow-up appointments.


Assuntos
Betacoronavirus/patogenicidade , Infecções por Coronavirus/imunologia , Neoplasias/imunologia , Peptidil Dipeptidase A/metabolismo , Pneumonia Viral/imunologia , Enzima de Conversão de Angiotensina 2 , Animais , Betacoronavirus/imunologia , COVID-19 , Infecções por Coronavirus/complicações , Citocinas/metabolismo , Humanos , Neoplasias/complicações , Nicotinamida Fosforribosiltransferase/metabolismo , Pandemias , Peptidil Dipeptidase A/imunologia , Pneumonia Viral/complicações , SARS-CoV-2
2.
Semin Immunol ; 35: 29-34, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29428698

RESUMO

Basophils, eosinophils and mast cells were first recognized by Paul Ehrlich in the late 19th century. These cells have common, but non-redundant roles, in the pathogenesis of allergic diseases and in the protection against parasites. Nevertheless, in virtue of their shared-adeptness to produce a huge variety of immunological mediators and express membrane-bound receptors, they are able to interact with immune and non-immune components of the tissue microenvironment, contributing to the regulation of tissue homeostasis and immune response while participating to further deregulation of tissues transforming into neoplasia.


Assuntos
Basófilos/imunologia , Eosinófilos/imunologia , Hipersensibilidade/imunologia , Mastócitos/imunologia , Neoplasias/imunologia , Animais , Transformação Celular Neoplásica , Microambiente Celular , Homeostase , Humanos
3.
ESMO Open ; 8(4): 101590, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37393630

RESUMO

BACKGROUND: Gene expression profiling (GEP)-based prognostic signatures are being rapidly integrated into clinical decision making for systemic management of breast cancer patients. However, GEP remains relatively underdeveloped for locoregional risk assessment. Yet, locoregional recurrence (LRR), especially early after surgery, is associated with poor survival. PATIENTS AND METHODS: GEP was carried out on two independent luminal-like breast cancer cohorts of patients developing early (≤5 years after surgery) or late (>5 years) LRR and used, by a training and testing approach, to build a gene signature able to intercept women at risk of developing early LRR. The GEP data of two in silico datasets and of a third independent cohort were used to explore its prognostic value. RESULTS: Analysis of the first two cohorts led to the identification of three genes, CSTB, CCDC91 and ITGB1, whose expression, derived by principal component analysis, generated a three-gene signature significantly associated with early LRR in both cohorts (P value <0.001 and 0.005, respectively), overcoming the discriminatory capability of age, hormone receptor status and therapy. Remarkably, the integration of the signature with these clinical variables led to an area under the curve of 0.878 [95% confidence interval (CI) 0.810-0.945]. In in silico datasets we found that the three-gene signature retained its association, showing higher values in the early relapsed patients. Moreover, in the third additional cohort, the signature significantly associated with relapse-free survival (hazard ratio 1.56, 95% CI 1.04-2.35). CONCLUSIONS: Our three-gene signature represents a new exploitable tool to aid treatment choice in patients with luminal-like breast cancer at risk of developing early recurrence.


Assuntos
Neoplasias da Mama , Feminino , Humanos , Neoplasias da Mama/genética , Neoplasias da Mama/tratamento farmacológico , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/tratamento farmacológico , Prognóstico , Transcriptoma , Medição de Risco
4.
J Exp Med ; 183(1): 317-22, 1996 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-8551239

RESUMO

The priming of an immune response against a major histocompatibility complex class I-restricted antigen expressed by nonhematopoietic cells involves the transfer of that antigen to a host bone marrow-derived antigen presenting cell (APC) for presentation to CD8+ T lymphocytes. Dendritic cells (DC), as bone marrow-derived APC, are first candidates for presentation of tumor-associated antigens (TAA). The aim of this study was to see whether DC are able to prime in vivo antigen-specific cytotoxic T lymphocytes after exposure to a soluble protein antigen in vitro. Lacking a well-defined murine TAA, we took advantage of beta-galactosidase (beta-gal)-transduced tumor cell lines as a model in which beta-gal operationally functions as TAA. For in vivo priming both a DC line, transduced or not transduced with the gene coding for murine GM-CSF, and fresh bone marrow-derived DC (bm-DC), loaded in vitro with soluble beta-gal, were used. Priming with either granulocyte macrophage colony-stimulating factor-transduced DC line or fresh bm-DC but not with untransduced DC line generated CTL able to lyse beta-gal-transfected target cells. Furthermore, GM-CSF was necessary for the DC line to efficiently present soluble beta-gal as an H-2Ld-restricted peptide to a beta-gal-specific CTL clone. Data also show that a long-lasting immunity against tumor challenge can be induced using beta-gal-pulsed bm-DC as vaccine. These results indicate that effector cells can be recruited and activated in vivo by antigen-pulsed DC, providing an efficient immune reaction against tumors.


Assuntos
Células Dendríticas/imunologia , Neoplasias Experimentais/terapia , Linfócitos T Citotóxicos/imunologia , Vacinação , Animais , Medula Óssea/imunologia , Células da Medula Óssea , Linfócitos T CD8-Positivos/imunologia , Células Clonais/imunologia , Feminino , Antígenos de Histocompatibilidade Classe I/imunologia , Esquemas de Imunização , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Fragmentos de Peptídeos/imunologia , Vacinas/imunologia , beta-Galactosidase/imunologia
5.
J Exp Med ; 190(1): 125-33, 1999 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-10429676

RESUMO

We transduced BALB/c-derived C-26 colon carcinoma cells with granulocyte/macrophage colony-stimulating factor (GM-CSF) and CD40 ligand (CD40L) genes to favor interaction of these cells with host dendritic cells (DCs) and, therefore, cross-priming. Cotransduced cells showed reduced tumorigenicity, and tumor take was followed by regression in some mice. In vivo tumors were heavily infiltrated with DCs that were isolated, phenotyped, and tested in vitro for stimulation of tumor-specific cytotoxic T lymphocytes (CTLs). BALB/c C-26 carcinoma cells express the endogenous murine leukemia virus (MuLV) env gene as a tumor-associated antigen. This antigen is shared among solid tumors of BALB/c and C57BL/6 mice and contains two epitopes, AH-1 and KSP, recognized in the context of major histocompatibility complex class I molecules H-2Ld and H-2K(b), respectively. DCs isolated from C-26/GM/CD40L tumors grown in (BALB/c x C57BL/6)F1 mice (H-2d x b) stimulated interferon gamma production by both anti-AH-1 and KSP CTLs, whereas tumor-infiltrating DCs (TIDCs) of BALB/c mice stimulated only anti-AH-1 CTLs. Furthermore, TIDCs primed naive mice for CTL activity as early as 2 d after injection into the footpad, whereas double-transduced tumor cells required at least 5 d for priming; this difference may reflect direct DC priming versus indirect tumor cell priming. Immunohistochemical staining indicated colocalization of DCs and apoptotic bodies in the tumors. These data indicate that DCs infiltrating tumors that produce GM-CSF and CD40L can capture cellular antigens, likely through uptake of apoptotic bodies, and mature in situ to a stage suitable for antigen presentation. Thus, tumor cell-based vaccines engineered to favor the interaction with host DCs can be considered.


Assuntos
Antígenos de Neoplasias/metabolismo , Células Dendríticas/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Glicoproteínas de Membrana/genética , Linfócitos T Citotóxicos/imunologia , Animais , Antígenos de Neoplasias/imunologia , Ligante de CD40 , Neoplasias do Colo/imunologia , Feminino , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos BALB C , Células Tumorais Cultivadas
6.
J Exp Med ; 188(1): 133-43, 1998 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-9653090

RESUMO

We analyzed the ability of interferon (IFN)-gamma knockout mice (GKO) to reject a colon carcinoma transduced with interleukin (IL)-12 genes (C26/IL-12). Although the absence of IFN-gamma impaired the early response and reduced the time to tumor onset in GKO mice, the overall tumor take rate was similar to that of BALB/c mice. In GKO mice, C26/IL-12 tumors had a reduced number of infiltrating leukocytes, especially CD8 and natural killer cells. Analysis of the tumor site, draining nodes, and spleens of GKO mice revealed reduced expression of IFN- inducible protein 10 and monokine induced by gamma-IFN. Despite these defects, GKO mice that rejected C26/IL-12 tumor, and mice that were primed in vivo with irradiated C26/IL-12 cells, showed the same cytotoxic T lymphocyte activity but higher production of granulocyte/macrophage colony-stimulating factor (GM-CSF) as compared with control BALB/c mice. Treatment with monoclonal antibodies against GM-CSF abrogated tumor regression in GKO but not in BALB/c mice. CD4 T lymphocytes, which proved unnecessary or suppressive during rejection of C26/IL-12 cells in BALB/c mice, were required for tumor rejection in GKO mice. CD4 T cell depletion was coupled with a decline in GM-CSF expression by lymphocytes infiltrating the tumors or in the draining nodes, and with the reduction and disappearance of granulocytes and CD8 T cells, respectively, in tumor nodules. These results suggest that GM-CSF can substitute for IFN-gamma in maintaining the CD8-polymorphonuclear leukocyte cross-talk that is a hallmark of tumor rejection.


Assuntos
Linfócitos T CD4-Positivos/fisiologia , Transformação Celular Neoplásica/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Interferon gama/genética , Interleucina-12/genética , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/fisiologia , Quimiocina CXCL10 , Quimiocinas CXC/análise , Neoplasias do Colo/metabolismo , Citotoxicidade Imunológica/genética , Citotoxicidade Imunológica/imunologia , Regulação Neoplásica da Expressão Gênica/genética , Imuno-Histoquímica , Hibridização In Situ , Interleucina-12/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Neoplasias Experimentais/genética , Nitritos/metabolismo , Linfócitos T Citotóxicos/metabolismo , Células Tumorais Cultivadas
7.
J Exp Med ; 173(4): 889-97, 1991 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-1706752

RESUMO

We have investigated the effect of granulocyte colony-stimulating factor (G-CSF) delivery at the site of tumor growth by transducing, via retroviral vector, the human (hu) G-CSF gene into the colon adenocarcinoma C-26 and assaying the ability of transduced cells to form tumors when injected into syngeneic mice. As a control, the same tumor cells were infected with retroviruses engineered to transduce an unrelated gene, the human nerve growth factor receptor, or carry the neomycin resistance gene only. Only cells transduced with the huG-CSF were unable to develop tumors, although huG-CSF was expressed and produced at low level as estimated by both RNA analysis and enzyme-linked immunosorbent assay, indicating that G-CSF can exert an antitumor effect at a physiological dose. Implication of G-CSF as mediator of tumor inhibition was proven by reversing the nontumorigenic phenotype of G-CSF-expressing cells with anti-huG-CSF monoclonal antibody injected at the tumor site. No tumors were formed by injecting C-26 infected cells into nu/nu mice, while neoplastic nodules appeared after injection into sublethally irradiated mice; such tumors, however, regressed when mice normalized their leukocyte counts after irradiation. Tumors were also formed after injection of a mixture of infected and uninfected C-26 cells, although critical delay in tumor formation occurred when infected cells were 10 times more represented in the mixture. Histological examination of tissues surrounding the site of injection showed infiltration of neutrophilic granulocytes, whose number correlated with that of G-CSF-expressing C-26 cells in the injected mixture. These results indicate that G-CSF may have a potent antitumoral activity when released, even at low doses, at the tumor site. The antitumoral effect is mediated by recruitment and targeting of neutrophilic granulocytes to G-CSF-releasing cells.


Assuntos
Adenocarcinoma/terapia , Neoplasias do Colo/terapia , Fator Estimulador de Colônias de Granulócitos/genética , Adenocarcinoma/imunologia , Adenocarcinoma/patologia , Animais , Neoplasias do Colo/imunologia , Neoplasias do Colo/patologia , Expressão Gênica , Terapia Genética , Fator Estimulador de Colônias de Granulócitos/fisiologia , Imunidade Celular , Técnicas Imunológicas , Camundongos , Camundongos Nus , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/patologia , Neoplasias Experimentais/terapia , Transfecção
8.
J Exp Med ; 178(1): 151-61, 1993 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-7686211

RESUMO

Using the murine colon adenocarcinoma C-26 cell line, engineered to release granulocyte colony-stimulating factor (G-CSF) (C-26/G-CSF), were studied the mechanisms responsible for inhibition of tumor take in syngeneic animals and of regression of an established tumor in sublethally irradiated mice injected with these cells. Immunocytochemistry and in situ hybridization, performed to characterize tumor-infiltrating leukocytes and their cytokine expression, respectively, indicated that polymorphonuclear leukocytes (PMN) were the major cells responsible for inhibition of tumor take and that they expressed mRNA for interleukin 1 alpha (IL-1 alpha), IL-1 beta, and tumor necrosis factor alpha (TNF-alpha). Expression of interferon gamma (IFN-gamma) and of IL-4 was undetectable, consistent with the absence of T lymphocytes at the site of tumor injection. In mice injected with C-26/G-CSF cells after 600-rad irradiation, the tumors grew to approximately 1.5 cm in 30 d, regressing completely thereafter in 70-80% of mice. During the growing phase, tumors were infiltrated first by PMN (between days 15 and 20), then by macrophages, and last by T lymphocytes. Both CD4+ and CD8+ T cells were present but only CD8 depletion significantly abrogated tumor regression. Depletion of PMN by the RB6-8C5 antigranulocytes monoclonal antibody reduced the number of T cells infiltrating the tumor and prevented tumor regression. In situ hybridization performed at the beginning of tumor regression revealed the presence of mRNA for IL-1 alpha, IL-1 beta, and TNF-alpha, but also the presence of cells, with lymphoid morphology, expressing IFN-gamma. Tumors from mice treated with recombinant IFN-gamma (between days 20 and 35) were rejected faster, whereas mice treated with antibodies to IFN-gamma (from day 20) died of progressive tumor. Cyclosporin A treatment (started at day 20) also abrogated tumor regression. These results indicate that inhibition of tumor take and regression in this model occurs through different mechanisms that involve PMN and PMN-T cell interactions, respectively, as well as a combination of cytokines that, for tumor regression, require IFN-gamma. Thus, gene transfer of a single cytokine gene such as G-CSF into tumor cells appears to be sufficient to trigger the cascade of cell interactions and cytokine production necessary to destroy a cancer nodule.


Assuntos
Adenocarcinoma/terapia , Neoplasias do Colo/terapia , Fator Estimulador de Colônias de Granulócitos/biossíntese , Interferon gama/fisiologia , Neutrófilos/imunologia , Linfócitos T/imunologia , Adenocarcinoma/imunologia , Adenocarcinoma/patologia , Animais , Anticorpos Monoclonais/imunologia , Comunicação Celular , Neoplasias do Colo/imunologia , Neoplasias do Colo/patologia , Ciclosporina/farmacologia , Fator Estimulador de Colônias de Granulócitos/genética , Camundongos , Camundongos Endogâmicos BALB C , Transfecção , Células Tumorais Cultivadas
9.
J Exp Med ; 194(9): 1195-205, 2001 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-11696586

RESUMO

Transgenic Balb/c mice expressing the transforming rat HER-2/neu oncogene develop early and multifocal mammary carcinomas. Within the first 5 months of life the tissue-specific expression of HER-2/neu causes a progression in all their 10 mammary glands from atypical hyperplasia to invasive carcinoma. It was previously observed that chronic administration of interleukin (IL)-12 increased tumor latency, but every mouse eventually succumbed to multiple carcinomas. A significant improvement in tumor prevention was sought by administering allogeneic mammary carcinoma cells expressing HER-2/neu combined with systemic IL-12. This treatment reduced tumor incidence by 90% and more than doubled mouse lifetime. For the maximum prevention p185(neu) antigen must be expressed by allogeneic cells. IL-12 treatment strongly increased the cell vaccine efficacy. The mammary glands of mice receiving the combined treatment displayed a markedly reduced epithelial cell proliferation, angiogenesis, and HER-2/neu expression, while the few hyperplastic foci were heavily infiltrated by granulocytes, macrophages, and CD8(+) lymphocytes. Specific anti-HER-2/neu antibodies were produced and a nonpolarized activation of CD4(+) and CD8(+) cells secreting IL-4 and interferon (IFN)-gamma were evident. A central role for IFN-gamma in the preventive effect was proven by the lack of efficacy of vaccination in IFN-gamma gene knockout HER-2/neu transgenic Balb/c mice. A possible requirement for IFN-gamma is related to its effect on antibody production, in particular on IgG2a and IgG2b subclasses, that were not induced in IFN-gamma knockout HER-2/neu mice. In conclusion, our data show that an allogeneic HER-2/neu-expressing cell vaccine combined with IL-12 systemic treatment can prevent the onset of genetically determined tumors.


Assuntos
Adjuvantes Imunológicos , Vacinas Anticâncer/imunologia , Interleucina-12/imunologia , Neoplasias Mamárias Experimentais/prevenção & controle , Receptor ErbB-2/fisiologia , Animais , Mama/patologia , Linfócitos T CD4-Positivos/imunologia , Transplante de Células , Feminino , Imunidade Celular/imunologia , Interferon gama/genética , Interferon gama/imunologia , Interleucina-12/administração & dosagem , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Transgênicos , Ratos , Receptor ErbB-2/genética , Transplante Homólogo , Células Tumorais Cultivadas , Vacinação/métodos
10.
J Exp Med ; 188(3): 589-96, 1998 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-9687535

RESUMO

The ability of interleukin (IL)-12 to prevent tumors when administered to individuals with a genetic risk of cancer was studied in two lines of transgenic mice expressing rat HER-2/neu oncogene in the mammary gland. Female BALB/c (H-2(d)) mice carrying the activated HER-2/ neu oncogene show no morphological abnormalities of the mammary gland until 3 wk of age. They then progress through atypical hyperplasia to in situ lobular carcinoma and at 33 wk of age all 10 mammary glands display invasive carcinomas. Adult FVB mice (H-2(q)) carrying the HER-2/neu protooncogene develop mammary carcinomas with a longer latency (38-49 wk) and a lower multiplicity (mean of 2.6 tumors/mice). Treatment with IL-12 (5 daily intraperitoneal injections, 1 wk on, 3 wk off; the first course with 50 ng IL-12/day, the second with 100 ng IL-12/day) begun at 2 wk of age in BALB/c mice and at 21 wk of age in FVB mice markedly delayed tumor onset and reduced tumor multiplicity. Analogous results were obtained in immunocompetent and permanently CD8(+) T lymphocyte-depleted mice. In both transgenic lines, tumor inhibition was associated with mammary infiltration of reactive cells, production of cytokines and inducible nitric oxide synthase, and reduction in microvessel number, in combination with a high degree of hemorrhagic necrosis.


Assuntos
Antineoplásicos/farmacologia , Carcinoma in Situ/prevenção & controle , Carcinoma Lobular/prevenção & controle , Interleucina-12/farmacologia , Neoplasias Mamárias Experimentais/prevenção & controle , Receptor ErbB-2/fisiologia , Animais , Antineoplásicos/imunologia , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Carcinoma in Situ/genética , Carcinoma in Situ/imunologia , Carcinoma Lobular/genética , Carcinoma Lobular/imunologia , Quimiocina CXCL10 , Quimiocina CXCL9 , Quimiocinas CXC/genética , Feminino , Interferon gama/imunologia , Interleucina-12/imunologia , Depleção Linfocítica , Masculino , Neoplasias Mamárias Experimentais/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Óxido Nítrico Sintase/biossíntese , Óxido Nítrico Sintase Tipo II , Ratos , Receptor ErbB-2/genética , Fator de Necrose Tumoral alfa/imunologia , Molécula 1 de Adesão de Célula Vascular/biossíntese
11.
Cancer Treat Rev ; 75: 39-51, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30954906

RESUMO

Immunotherapy has dramatically changed the therapeutic scenario in treatment naïve advanced non-small cell lung cancer (NSCLC). While single agent pembrolizumab has become the standard therapy in patients with PD-L1 expression on tumor cells ≥ 50%, the combination of pembrolizumab or atezolizumab and platinum-based chemotherapy has emerged as an effective first line treatment regardless of PD-L1 expression both in squamous and non-squamous NSCLC without oncogenic drivers. Furthermore, double immune checkpoint inhibition has shown promising results in treatment naïve patients with high tumor mutational burden (TMB). Of note, the presence of both negative PD-L1 expression and low TMB may identify a subgroup of patients who has little benefit from immunotherapy combinations and for whom the best treatment option may still be platinum-based chemotherapy. To date, first-line single agent immune checkpoint blockade has demonstrated limited activity in EGFR mutated NSCLC and the combination of immunotherapy and targeted agents has raised safety concerns in both EGFR and ALK positive NSCLC patients. Finally, in EGFR mutated or ALK rearranged NSCLC, atezolizumab in combination with platinum-based chemotherapy and bevacizumab is emerging as a potential treatment option upon progression to first line tyrosine kinase inhibitors.


Assuntos
Antineoplásicos/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/imunologia , Carcinoma Pulmonar de Células não Pequenas/terapia , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/terapia , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados/uso terapêutico , Antígeno B7-H1/metabolismo , Bevacizumab/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Humanos , Fatores Imunológicos/metabolismo , Imunoterapia/métodos , Neoplasias Pulmonares/metabolismo
12.
Mol Cell Biol ; 16(9): 4842-51, 1996 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8756643

RESUMO

Homeobox (HOX) genes control axial specification during mammalian development and also regulate skin morphogenesis. Although selected HOX genes are variably expressed in leukemias and kidney and colon cancer cell lines, their relationship with the neoplastic phenotype remains unclear. In both normal development and neoplastic transformation, HOX target genes are largely unknown. We investigated the expression and function of HOXB cluster genes in human melanoma. The HOXB7 gene was constitutively expressed in all 25 melanoma cell lines and analyzed under both normal and serum-starved conditions, as well as in in vivo primary and metastatic melanoma cells; conversely, HOXB7 was expressed in proliferating but not quiescent normal melanocytes. Treatment of melanoma cell lines with antisense oligomers targeting HOXB7 mRNA markedly inhibited cell proliferation and specifically abolished expression of basic fibroblast growth factor (bFGF) mRNA. Band shift and cotransfection experiments showed that HOXB7 directly transactivates the hFGF gene through one out of five putative homeodomain binding sites present in its promoter. These novel findings indicate a key role for constitutive HOXB7 expression in melanoma cell proliferation via bFGF. The results also raise the possibility that growth factor genes are critical HOX target genes in other developmental and/or neoplastic cell systems.


Assuntos
Fator 2 de Crescimento de Fibroblastos/metabolismo , Regulação Neoplásica da Expressão Gênica , Proteínas de Homeodomínio/fisiologia , Melanoma/metabolismo , Proteínas de Neoplasias/metabolismo , Elementos Antissenso (Genética) , Sequência de Bases , Meios de Cultura Livres de Soro , Genes Homeobox , Células HeLa , Humanos , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , RNA Mensageiro/biossíntese , Proteínas Recombinantes de Fusão/biossíntese , Transfecção , Células Tumorais Cultivadas
13.
Cytokine Growth Factor Rev ; 38: 1-9, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29029813

RESUMO

The seventh Edition of "Innovative Therapy, Monoclonal Antibodies and Beyond" Meeting took place in Milan, Italy, on January 27, 2017. The two sessions of the meeting were focused on: 1) Preclinical assays and novel biotargets; and 2) monoclonal antibodies, cell therapies and targeted molecules. Between these two sessions, a lecture entitled "HLA-antigens modulation and response to immune checkpoint inhibitor immunotherapy" was also presented. Despite the impressive successes in cancer immunotherapy in recent years, the response to immune based interventions occurs only in a minority of patients (∼20%). Several basic and translational mechanisms of resistance to immune checkpoint blockers (ICBs) were discussed during the meeting: 1. the impact of tumor microenvironment on the activity of immune system; 2. strategies to inhibit the cross-talk between extracellular matrix and myeloid-derived suppressor cells (MDSC) in the preclinical setting; 3. microRNA expression as a biomarker and as a target of therapy in non-small cell lung cancer (NSCLC); 4. the significance of complement activation pathways in response to immune checkpoint inhibitors; 5. the immunosuppressive activity of the microbiota by inducing IL-17 producing cells; and 6. modulation of HLA antigens as possible markers of response to ICB therapy. In order to overcome the deficiency in active anti-tumor T cells, several clinically applicable combination strategies were also discussed: 1. strategies to enhance the anticancer effects of immunogenic cell death inducing-chemotherapy; 2. the use of CAR T-cells in solid tumors; 3. the use of combination strategies involving oncolytic viruses and ICBs; 4. combinations of new ICBs with anti-PD-1/CTLA-4 therapy; and 4. combinations of targeted therapies and ICBs in melanoma. Overall, this conference emphasized the many novel strategies that are being investigated to improve the overall patient response to cancer immunotherapy. Optimization of biomarkers to accurately select patients who will respond to immunotherapy, coupled with combination strategies to improve long term patient survival remain critical challenges in the immuno-oncology field.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Imunoterapia , Neoplasias/terapia , Animais , Humanos , Neoplasias/tratamento farmacológico
14.
J Natl Cancer Inst ; 89(14): 1049-58, 1997 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-9230887

RESUMO

BACKGROUND: Numerous animal model studies have examined the ability of genetically engineered tumor cells to release cytokines and to elicit an immune memory against the parental tumor. Often only a single cytokine is studied, and few comparative studies have been conducted. PURPOSE: We evaluated the antitumor efficacy of adenocarcinoma cells engineered to release interleukin (IL)-12 in a mouse model system. The efficacy of this cytokine was compared with that of other cytokines released by engineered adenocarcinoma cells and that of exogenous IL-12 administered both locally and intraperitoneally. METHODS: BALB/cAnCr mice were inoculated with syngeneic parental mammary adenocarcinoma (TSA) cells in quantities sufficient to lead to tumors in all inoculated mice. TSA cells engineered to release IL-12 (TSA-IL12) were also injected into normal and selectively immunosuppressed BALB/cAnCr mice. Tumor incidence, growth, and rejection patterns were evaluated by the measurement of neoplastic masses and by the study of the histologic and ultrastructural features of the tumor site. The effects of local or intraperitoneal administration of recombinant IL-12 (rIL-12) on tumor-bearing animals were also studied. RESULTS: Most mice rejected TSA-IL12 cells through a CD8-positive, T-lymphocyte-dependent reaction associated with macrophage infiltration, vessel damage, and necrosis. The systemic immunity of mice that had rejected TSA-IL12 cells to a subsequent challenge with parental TSA cells was less efficient than that elicited by TSA cells engineered to release IL-4 or IL-10 but equivalent to that elicited by TSA cells engineered to release IL-2, IL-7, and interferon alfa. Compared with TSA cells engineered to produce other cytokines, TSA-IL12 cells were the most efficient in curing mice with established TSA tumors; injection of 0.1 million proliferating cells contralaterally to the tumor growth area cured five of 15 mice bearing 1-day-old tumors; injection of the same dose of proliferating cells into the tumor growth area cured two of 20 tumor-bearing mice. However, two 5-day courses with a nontoxic dose (0.1 microgram) of rIL-12 given intraperitoneally cured a similar proportion of these animals (six of 20). Only two of 20 mice with 7-day-old TSA tumors were cured by vaccination with proliferating TSA-IL12 cells, whereas 24 of 30 mice with such tumors were cured by intraperitoneal administration of rIL-12. CONCLUSIONS: TSA cells engineered to release IL-12 are rejected by most mice; the ensuing immune memory for TSA parental cells, however, was less efficient than that elicited by proliferating TSA cells engineered to release other cytokines (e.g., IL-4, IL-10, and possibly interferon gamma). The immune reaction elicited by TSA-IL12 cells was the most efficient in curing mice with established TSA tumors; notably though, the same or a better cure rate was obtained with rIL-12 given intraperitoneally.


Assuntos
Adenocarcinoma/tratamento farmacológico , Interleucina-12/farmacologia , Interleucina-12/fisiologia , Neoplasias Mamárias Experimentais/tratamento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/patologia , Animais , Modelos Animais de Doenças , Feminino , Infusões Parenterais , Injeções Intralesionais , Interleucina-12/administração & dosagem , Interleucina-12/biossíntese , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/farmacologia , Transdução Genética
15.
Cancer Res ; 51(11): 2897-901, 1991 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-2032228

RESUMO

Steady-state mRNA levels of the protooncogene c-myb were measured by Northern blot analysis in the human colon carcinoma cell lines LoVo, the doxorubicin-resistant derivative LoVo/Dx, Colo 205, and HT 29. Overexpression of c-myb mRNA was detected in the Colo 205 cell line, probably because of gene amplification, while in human HT 29 cells c-myb was not expressed at a detectable level. Comparison between LoVo and LoVo/Dx cell lines showed that c-myb mRNA levels were much higher in the doxorubicin-resistant derivative than in the parental line. c-myb antisense oligodeoxynucleotides inhibited cell proliferation only in the cell lines with detectable mRNA c-myb (LoVo, LoVo/DX, and Colo 205). The dose of antisense exerting inhibitory effect was related to the levels of c-myb mRNA expression. Inhibition of c-myb expression in antisense-treated LoVo/DX cells was demonstrated by the reverse transcriptase polymerase chain reaction technique. LoVo/Dx cells were induced to differentiate by treatment with dimethylformamide to determine whether down-regulation of c-myb expression would accompany the process of differentiation. During the treatment with dimethylformamide the expression of c-myb decreased in parallel with the reduction of cell growth, while terminal differentiation of these cells was associated with changes in the expression of carcinoembryonic antigen and laminin receptor genes. Our findings demonstrate that the expression of c-myb is important for the proliferation of colon carcinoma cell lines and suggest that the role of this protooncogene is not restricted to cells of hematopoietic origin but is more general than previously thought.


Assuntos
Adenocarcinoma/genética , Neoplasias do Colo/genética , Regulação Neoplásica da Expressão Gênica/genética , Oligonucleotídeos Antissenso/farmacologia , Oncogenes/efeitos dos fármacos , RNA Mensageiro/análise , RNA Neoplásico/análise , Divisão Celular/efeitos dos fármacos , Divisão Celular/genética , Dimetilformamida/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Reação em Cadeia da Polimerase , RNA Mensageiro/genética , RNA Neoplásico/genética , Células Tumorais Cultivadas
16.
Cancer Res ; 60(10): 2571-5, 2000 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-10825122

RESUMO

Immunotherapy applied to patients with established tumors rarely leads to an objective response, whereas patients apparently free from disease after conventional treatment and at risk of recurrence are beginning to receive vaccination. New classes of patients or not-yet patients are those with a high genetic or environmental risk of developing cancer. They may draw benefit from a "soft" treatment such as vaccination. This overview discusses the prospects of immune stimulation as a means of cancer prevention by inducing various forms of nonspecific or even specific immunity. Attainment of this goal provides the rationale and motivation for embarking on such a new and potentially rewarding enterprise.


Assuntos
Imunização , Neoplasias/prevenção & controle , Humanos , Neoplasias/imunologia
17.
Cancer Res ; 60(14): 3777-81, 2000 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-10919650

RESUMO

An approach to stimulating an immune response against tumors is to transduce tumor cells with bacterial genes, which represent a "danger signal" and can induce a wide immune response. Mycobacterium tuberculosis genes and their encoded proteins play a pivotal role in linking innate and cell-mediated adaptive immunity and represent ideal candidates as immune adjuvants for tumor vaccines. The efficacy of a cancer vaccine, obtained by transduction of a mammary tumor cell line with the M. tuberculosis Ag38 gene, was investigated in female mice transgenically expressing the rat HER-2/neu proto-oncogene. These mice spontaneously develop stochastic mammary tumors after a long latency period. The onset of spontaneous mammary tumors was significantly delayed in mice vaccinated with Ag38-transduced cells but not in mice vaccinated with nontransduced cells as compared with untreated mice. Protection from spontaneous tumor development was increased when mice were vaccinated with the mycobacterium gene-transduced vaccine plus a systemic administration of interleukin 12 (IL-12) at a low dose. Mice vaccinated with nontransduced cells plus IL-12 developed tumors, with only a slight delay in tumor appearance as compared with the control group. Lymphocytes obtained from lymph nodes of mice vaccinated with transduced cells secreted high levels of IFN-gamma. CD3+CD8+ spleen cells derived from these mice responded to the tumor with IFN-gamma production. These data indicate the efficacy of a short-term protocol of vaccinations exploiting the adjuvant potency of a M. tuberculosis gene and low doses of IL-12 in a model of stochastic development of mammary tumors. This adjuvant approach may represent a promising immunotherapeutic strategy for cancer immunization.


Assuntos
Antígenos de Bactérias/genética , Vacinas Anticâncer , Interleucina-12/farmacologia , Lipoproteínas/genética , Receptor ErbB-2/genética , Transdução Genética , Adjuvantes Imunológicos/farmacologia , Fatores Etários , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Divisão Celular/efeitos dos fármacos , Divisão Celular/genética , Separação Celular , Células Cultivadas , Citocinas/biossíntese , Relação Dose-Resposta a Droga , Feminino , Citometria de Fluxo , Técnicas de Transferência de Genes , Interferon gama/biossíntese , Interferon gama/farmacologia , Interleucina-4/biossíntese , Linfonodos/metabolismo , Neoplasias Mamárias Experimentais/induzido quimicamente , Neoplasias Mamárias Experimentais/genética , Camundongos , Camundongos Transgênicos , Ratos , Proteínas Recombinantes/farmacologia , Baço/metabolismo , Células Tumorais Cultivadas
18.
Cancer Res ; 55(10): 2200-5, 1995 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-7743524

RESUMO

The cytokines interleukin 6 (IL-6) and oncostatin M are able to inhibit the growth of cell lines obtained from early but not advanced melanomas. Resistant cell lines have frequently been found to produce IL-6. Acquisition of IL-6 resistance and the relationship between resistance and endogenous IL-6 production are poorly defined phenomena. We have characterized a panel of melanoma cell lines for susceptibility to IL-6 and oncostatin M and have generated lines that acquired resistance to IL-6 by IL-6 cDNA transduction. These lines retained the previous oncostatin M sensitivity, suggesting that the alpha chain of IL-6 receptor (IL-6R alpha) is involved in the acquisition of resistance. In fact transduced cells lost the ability to bind 125I-IL-6 and to release soluble IL-6R alpha in culture. Moreover, addition of soluble recombinant IL-6R alpha were able to restore IL-6 sensitivity in association with IL-6 production. On the contrary, naturally IL-6 resistant melanoma cell lines were not inhibited by treatment with recombinant soluble IL-6R alpha in association with endogenously produced or recombinant IL-6. These results demonstrate that down-regulation of IL-6 receptor is only one of different mechanisms that are responsible of IL-6 resistance in melanoma cells.


Assuntos
Regulação para Baixo , Interleucina-6/metabolismo , Melanoma/metabolismo , Peptídeos/metabolismo , Receptores de Interleucina/metabolismo , Divisão Celular/efeitos dos fármacos , Resistência a Medicamentos/genética , Humanos , Melanoma/genética , Melanoma/patologia , Oncostatina M , Receptores de Interleucina-6 , Transfecção , Células Tumorais Cultivadas
19.
Cancer Res ; 59(2): 414-21, 1999 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-9927056

RESUMO

The antitumor activity of recombinant murine interleukin-12 (rIL-12) is documented by a large set of data from numerous mouse models. Because the cellular and molecular mechanisms by which rIL-12 impairs tumor growth are still not fully defined, we compared the effects of local and systemic rIL-12 administration in mice harboring an invasive 7-day-old moderately differentiated and spontaneously metastasizing mammary adenocarcinoma (TSA). Whereas the immune events elicited via the two routes of rIL-12 administration seem to be the same, systemic rIL-12 is markedly more effective; tumor destruction is dependent on a prompt antitumor response resulting from the cooperation of several subsets of reactive cells. The reactions that seem to play a key role are: (a) indirect inhibition of angiogenesis by secondary cytokines (mainly IFN-gamma) and third-level chemokines (inducible protein 10 and monokine induced by IFN-gamma); (b) systemic activation of leukocyte subsets capable of producing proinflammatory cytokines, CTLs, and antitumor antibodies; and (c) destruction of tumor vessels by polymorphonuclear cells. The markedly higher efficacy of systemic rIL-12 seems to rest on its ability to recruit these systemic reactions more quickly and efficiently than local rIL-12.


Assuntos
Interleucina-12/uso terapêutico , Neoplasias Mamárias Experimentais/terapia , Animais , Anticorpos Antineoplásicos/sangue , Citocinas/genética , Citocinas/metabolismo , Feminino , Imuno-Histoquímica , Interleucina-12/administração & dosagem , Neoplasias Mamárias Experimentais/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Metástase Neoplásica , Neutrófilos/fisiologia , RNA Mensageiro/análise , Proteínas Recombinantes/uso terapêutico , Linfócitos T Citotóxicos/imunologia
20.
Cancer Res ; 56(11): 2531-4, 1996 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-8653692

RESUMO

The C26 colon carcinoma is resistant to systemic recombinant interleukin 12 (rIL-12) therapy. Transduction of C26 with genes encoding the two subunits of murine IL-12 to release 30-80 pg/ml resulted in delayed tumor onset after injection of 5 x 10(4) cells into syngeneic BALB/c mice and in 40% tumor regression after injection into CD4-depleted mice. Here, we analyzed the activity of rIL-12 (1 microgram/day) against C26 grown into CD4-depleted mice. Like in mice given injections of interleukin 12 (IL-12) gene-transduced C26 cells, depletion of CD4+ cells led to tumor regression in 6 of 14 mice, and immumocytochemical characterization of tumor-infiltrating leukocytes showed abundant infiltration by CD8+ T cells and asialoGM1+ natural killer cells, which were scanty in tumors from nondepleted mice. On the basis of the percentage of tumor regression and leukocyte infiltration we can conclude that, in the C26 system, systemic rIL-12 (1 pmicrogramg/day) produces the same results as 30-80 pg/ml IL-12 released at the tumor site. A new polycistronic retroviral vector was then used to increase the amount of IL-12 produced by C26-transduced cells. C26 cells releasing 5 ng/ml IL-12, nearly 100 times more than the above-mentioned transduced cells, were tumorigenic in less than 50% of the mice given injections of 5 x 10(4) cells. In mice given injections of 5 x 10(5) cells, an initial tumor take of 100% followed by a complete tumor regression. Tumor regression was associated with infiltration of CD8+ and asialoGM1+ cells, and mice that remained tumor free were immune to a rechallenge of nontransduced C26 cells. The results indicate that the amount of IL-12 made available at the tumor site may determine both the type and number of infiltrating leukocytes and the events leading to tumor regression as well as it may overcame host immunosuppression.


Assuntos
Adenocarcinoma/imunologia , Neoplasias do Colo/imunologia , Interleucina-12/administração & dosagem , Adenocarcinoma/patologia , Animais , Sequência de Bases , Linfócitos T CD4-Positivos/imunologia , Neoplasias do Colo/patologia , Primers do DNA/química , Memória Imunológica , Interleucina-12/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Proteínas Recombinantes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA